Microglia in Health and Disease - …cshperspectives.cshlp.org/content/8/1/a020560.full.pdf ·...

16
Microglia in Health and Disease Richard M. Ransohoff 1 and Joseph El Khoury 2 1 Neuroinflammation Research Center, Department of Neurosciences, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195 2 Division of Infectious Diseases and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129 Correspondence: [email protected]; [email protected] Microglia, the major myeloid cells of the central nervous system (CNS) are implicated in physiologic processes and in the pathogenesis of several CNS disorders. Since their initial description early in the 20th century, our ability to identify and isolate microglia has sig- nificantly improved and new research is providing insight into the functions of these cells in sickness and in health. Here, we review recent advances in our understanding of the role of microglia in physiological and pathological processes of the CNS with a focus on multiple sclerosis and Alzheimer’s disease. Because of the prominent roles CX3CR1 and its ligand fractalkine played in bringing about these advances, we discuss the physiological and pathological roles of microglia as viewed from the CX3CR1–fractalkine perspective, pro- viding a unique viewpoint. Based on the most recent studies of molecular profiling of microg- lia, we also propose a molecular and functional definition of microglia that incorporates the properties attributed to these cells in recent years. A BRIEF HISTORY OF MICROGLIA M icroglia, the major myeloid cells of the cen- tral nervous system (CNS), have long been implicated in physiologic and pathologic pro- cesses. The 20th century brought several major breakthroughs in our ability to identify and iso- late these cells. In the past decade, new research has emerged providing evidence and insight into the functions of these cells in sickness and in health. The first breakthroughs in research on mi- crogliawere presented in a series of publications, nearly a century ago, by Pı´o del Rı ´o-Hortega. In an article published in 1918, del Rı´o-Hortega described a method for staining microglia and was therefore able to distinguish these cells from other neighboring cells of the neural milieu (del Rı´o-Hortega 1918). In a subsequent article pub- lished the following year, del Rı´o-Hortega de- scribed microglia as the “third element” in the CNS, he also emphasized their phagocytic ca- pacity (del Rı ´o-Hortega 1919). Additional work performed by del Rı ´o-Hortega indirectly de- scribed microglial plasticity and discussed the ability of these cells to change in response to stimuli (del Rı ´o-Hortega 1920). He further went on to describe the regional distribution of microglia and their heterogeneity (del Rı´o- Hortega 1921). Since then, scientists investigat- ing microglia using progressively sophisticated technologies have largely confirmed and built on del Rı´o-Hortega’s initial observations. Editors: Ben A. Barres, Marc R. Freeman, and Beth Stevens Additional Perspectives on Glia available at www.cshperspectives.org Copyright # 2016 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a020560 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560 1 on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/ Downloaded from

Transcript of Microglia in Health and Disease - …cshperspectives.cshlp.org/content/8/1/a020560.full.pdf ·...

Microglia in Health and Disease

Richard M. Ransohoff1 and Joseph El Khoury2

1Neuroinflammation Research Center, Department of Neurosciences, Cleveland Clinic Lerner ResearchInstitute, Cleveland, Ohio 44195

2Division of Infectious Diseases and Center for Immunology and Inflammatory Diseases, MassachusettsGeneral Hospital, Harvard Medical School, Charlestown, Massachusetts 02129

Correspondence: [email protected]; [email protected]

Microglia, the major myeloid cells of the central nervous system (CNS) are implicated inphysiologic processes and in the pathogenesis of several CNS disorders. Since their initialdescription early in the 20th century, our ability to identify and isolate microglia has sig-nificantly improved and new research is providing insight into the functions of these cells insickness and in health. Here, we review recent advances in our understanding of the role ofmicroglia in physiological and pathological processes of the CNS with a focus on multiplesclerosis and Alzheimer’s disease. Because of the prominent roles CX3CR1 and its ligandfractalkine played in bringing about these advances, we discuss the physiological andpathological roles of microglia as viewed from the CX3CR1–fractalkine perspective, pro-viding a unique viewpoint. Based on the most recent studies of molecular profiling of microg-lia, we also propose a molecular and functional definition of microglia that incorporates theproperties attributed to these cells in recent years.

A BRIEF HISTORY OF MICROGLIA

Microglia, the major myeloid cells of the cen-tral nervous system (CNS), have long been

implicated in physiologic and pathologic pro-cesses. The 20th century brought several majorbreakthroughs in our ability to identify and iso-late these cells. In the past decade, new researchhas emerged providing evidence and insightinto the functions of these cells in sickness andin health.

The first breakthroughs in research on mi-crogliawere presented in a series of publications,nearly a century ago, by Pıo del Rıo-Hortega.In an article published in 1918, del Rıo-Hortegadescribed a method for staining microglia andwas therefore able to distinguish these cells from

other neighboring cells of the neural milieu (delRıo-Hortega 1918). In a subsequent article pub-lished the following year, del Rıo-Hortega de-scribed microglia as the “third element” in theCNS, he also emphasized their phagocytic ca-pacity (del Rıo-Hortega 1919). Additional workperformed by del Rıo-Hortega indirectly de-scribed microglial plasticity and discussedthe ability of these cells to change in responseto stimuli (del Rıo-Hortega 1920). He furtherwent on to describe the regional distributionof microglia and their heterogeneity (del Rıo-Hortega 1921). Since then, scientists investigat-ing microglia using progressively sophisticatedtechnologies have largely confirmed and builton del Rıo-Hortega’s initial observations.

Editors: Ben A. Barres, Marc R. Freeman, and Beth Stevens

Additional Perspectives on Glia available at www.cshperspectives.org

Copyright # 2016 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a020560

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560

1

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

After del Rıo-Hortega’s initial findings, ittook another 50 years for the second wave ofbreakthroughs in microglia research to occur.Development of antibodies to various innateimmune cell proteins identified markers for mi-croglia expressed in situ such as F4/80 (Humeet al. 1983), Fc receptors, and complement re-ceptor 3 (Perry et al. 1985). Subsequently, Giu-lian and Baker (1986) described a novel methodto isolate and culture microglia from neonatalrodent brains. The ability to culture these cellsled to a large number of in vitro investigations todefine their functions. Cultured microglia werefound to be capable of phagocytosis and lyso-zyme production (Zucker-Franklin et al. 1987),and respond to purinergic stimulation (Walzet al. 1993) and to b-amyloid (Meda et al.1995; El Khoury et al. 1996). This approachalso led to identification of novel markers forthese cells such as Iba1 (Imai et al. 1996).

The availability of new markers for microg-lia provided tools to begin to investigate the roleof these cells in various CNS disorders. Presenceof these cells in senile plaques in Alzheimer’sdisease (AD) was confirmed using specificmonoclonal antibodies to a1 chymotrypsin, ly-sozyme (Rozemuller et al. 1986), and HLA-DR(McGeer et al. 1987). Microglia-expressingMHC II antigens were also identified in multiplesclerosis (MS) brains (Cuzner et al. 1988).

The third era of breakthroughs in mi-croglia research started with the generation ofa transgenic knockin mouse expressing greenfluorescent protein (GFP) driven by the pro-moter for the chemokine receptor CX3CR1(Jung et al. 2000). Because CX3CR1 is exclusive-ly expressed on microglia in the CNS, GFP wasexpressed only in these cells, allowing investi-gators to visualize microglia without the needfor immunohistochemistry or special staining.This mouse line ushered in a new line of inves-tigations exploring the roles of microglia in var-ious physiological and pathological processes.

The ability to visualize microglia withoutthe need for immunolabeling and the devel-opment of novel two-photon in vivo imagingmethodologies provided unprecedented viewsinto the role of these cells in vivo. In a landmarkpublication, Nimmerjahn et al. (2005) showed

that the morphologically “resting” microgliaare highly active, continually surveying theirmicroenvironment with extremely motile pro-cesses and protrusions; they also showed thatfocal injury to the brain provoked immediateand focal activation of microglia, switchingtheir behavior from patrolling to shielding ofthe injured site. At the same time, Davalos andcolleagues (2005) reported identical findingsand showed further that the rapid process–extension response to laser lesion was mediatedby ATP binding to the purinergic receptorP2ry12 (Haynes et al. 2006). Additional stud-ies, discussed in the following pages, used theCX3CR1 GFP mice (or improved transgenicversions of these mice) and two-photon invivo imaging to further explore the role of mi-croglia in synaptic pruning and remodeling,and in various disease models including ex-perimental autoimmune encephalitis (EAE),a model of MS, mouse models of amyloid de-position and tauopathies in AD, and models ofischemic injury.

The fourth era of breakthroughs in microg-lia research began when methods to isolate mi-croglia from adult mice were developed (Hick-man et al. 2008), leading to several landmarkfindings in 2013. With the increased recogni-tion of the important roles microglia couldplay in physiological and pathological process-es, and because of the multiple similarities anddifferences between these cells and other mono-nuclear phagocytes, it became crucial to de-velop a more accurate definition of microgliathat distinguishes them from other cells in theCNS as well as other mononuclear phagocytessuch as monocytes and macrophages. Over thepast year, using methodologies such as directRNA sequencing (RNASeq) (Chiu et al. 2013;Hickman et al. 2013), and microarrays (Beutneret al. 2013; Butovsky et al. 2014), four groupsindependently identified expression signaturesof microglia that help define these cells and de-termine their unique transcriptional character-istics.

One further notable advance in under-standing microglia came gradually over de-cades, so cannot conveniently be summarizedin a single breakthrough. Microglial ontogeny

R.M. Ransohoff and J. El Khoury

2 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

was implied by del Rıo-Hortega as being mes-enchymal. However, unequivocal evidence forthis proposal was not forthcoming until 1996,when Maki and coworkers (McKercher et al.1996) showed that mice lacking transcriptionfactor PU.1 were devoid of microglia. In thesame time frame, Alliot and colleagues (1991,1999) conducted a series of descriptive studiesand proposed that microglia originate in theyolk sac, enter brain rudiment after E8 in rodentsand establish the adult microglial populationthrough vigorous in situ proliferation. Ajamiand colleagues followed in 2007, by showingthat microglia self-renew in the CNS without ablood-monocyte contribution. In 2010, Gin-houx and colleagues (2010) performed a defin-itive fate mapping confirming Pessac’s hypoth-esis. Two years later, Geissmann (Schulz et al.2012) showed the c-myb independence of mi-croglia and other tissue macrophages, conclu-sively establishing that microglial maintenancedid not rely on progeny of hematopoietic stemcells. With these insights came the ability toidentify microglial survival or maintenance fac-tors including colonystimulating factor (CSF)-1and interleukin (IL)-34 (Greteret al. 2012; Wanget al. 2012), both produced in the CNS by neu-rons during development and both ligands forthe CSF-1 receptor. Additionally, transcriptionfactors such as interferon regulatory factor(IRF)-8 could be shown essential for microglialphenotypes by Kierdorf and Prinz (2013).

In the following pages, we will review re-cent advances in our understanding of the roleof microglia in physiological and pathologi-cal processes of the CNS. Because of the promi-nent roles CX3CR1 and its ligand fractalkineplayed in bringing about these advances, andbecause fractalkine-induced, CX3CR1-mediat-ed signaling appears to regulate microglialbehavior in a number of CNS disorders in-cluding AD and MS, we will discuss the physio-logical roles of microglia as viewed from theCX3CR1–fractalkine perspective, providing aunique viewpoint. Based on the most recentstudies of molecular profiling of microglia, wewill also propose a molecularand functional def-inition of microglia that incorporates the prop-erties attributed to these cells in recent years.

PHYSIOLOGICAL ROLES OF MICROGLIAVIEWED FROM THE FRACTALKINE-CX3CR1PERSPECTIVE

Fractalkine (CX3CL1) and its receptor(CX3CR1) have assumed a prominent andunique role in CNS development, homeostasis,and disease within a relatively short time. Themost salient and well-established neurobiologi-cal role of this ligand/receptor pair is definedin large part by their cellular sources. Fractal-kine is made by neurons and the receptor isexpressed by microglia. Therefore, fractalkine/receptor interactions constitute a neuron–mi-croglial signaling system. Simply stated, effectsof neuronal fractalkine mediated by microglialfractalkine receptor provide a capsule view ofneuron–microglial cross talk throughout devel-opment, physiology, aging, and disease.

Early History of Fractalkine and Its Receptor

Fractalkine is a chemokine, albeit unusual. Che-mokines were identified by virtue of mediat-ing leukocyte subtype-specific chemotaxis invitro in 1987 (Ransohoff 2005). Four years lat-er, cloning of the first chemokine receptorcDNA (Horuk 1994) showed that chemokinereceptors belong to the superfamily of G pro-tein–coupled receptors (GPCRs), and subse-quent work showed that chemokine receptorscomprise a GPCR subclass signaling throughGai. Genomic analysis assigned most chemo-kines and chemokine receptors to a restrictedset of chromosomal loci, indicating their statusas a coherent genetic and functional family(Charo and Ransohoff 2006). At present, hu-mans and mice possess approximately 50 che-mokines and 20 chemokine receptors. Two largestructurally defined families, termed CC andCXC chemokines, include more than 90% ofall chemokines. In primary structures of CCchemokines (CCL[ligand]1–28), two position-ally conserved cysteines are side by side, whereasCXC (CXCL1–16) chemokines possess an in-tervening residue between the cysteines. CCchemokines signal almost without exceptiononly to CC chemokine receptors (CCR[recep-tor]1–12), and CXC chemokines show a similar

Microglia in Health and Disease

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560 3

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

preference for a corresponding set of CXC che-mokine receptors (CXCR1-7) (Rot and von An-drian 2004). Ligand–receptor relationships inthe chemokine universe are complex. Chemo-kine receptors can respond to as many as 10ligands, whereas individual chemokines cansignal to as many as three receptors.

Virtually all chemokines are small (10–14 kDa) secreted peptides, transcribed rapidlyand abundantly on demand with short-livedmessages and quickly cleared by degradationof protein and message as well as receptor-me-diated uptake and disposal of the peptides (Car-dona et al. 2008).

From their initial identification until 1998,chemokines were viewed mainly in the contextof inflammation and immunity because of theirpotent, selective action toward leukocyte popu-lations. Deletion of chemokine receptor CXCR4from the mouse genome upended this view ofchemokine physiology, showing prominent de-fects in multiple tissues, including abnormallocalization of numerous neuronal populations(Ma et al. 1998; Zou et al. 1998). These resultsushered in an era of chemokine neurobiology,with fractalkine constituting a conspicuous ele-ment.

Fractalkine was identified by molecularcloning in 1997 (Bazan et al. 1997) and showedtwo unusual features. First, the primary trans-lation product was not secreted and showed asingle-pass transmembrane region, long mucinstalk, and amino-terminal chemokine ectodo-main. Second, the canonical cysteine residuesshowed a CXXXC (CX3C) motif. In 2014, frac-talkine and fractalkine receptor remain the solemembers of their chemokine family. No otherCX3C chemokines have been discovered andonly one additional membrane tethered chemo-kine (CXCL16) has emerged. Fractalkine/frac-talkine receptor appear, for practical purposes,to constitute a monogamous ligand/receptorsystem. One other signaling CX3CR1 ligandwas reported (CCL26) but follow-up has beenscant so its biological significance is uncertain(Nakayama et al. 2010).

Fractalkine’s unusual name deserves a com-ment. Apparently the lead author was remindedof fractals, in which a repeating subunit recapit-

ulates the whole, by fractalkine’s primary struc-tural characteristics.

Fractalkine was quickly shown to be ex-pressed by endothelial cells, regulated in partby inflammatory cytokines. Initial studiesshowed that fractalkine signaled to receptor-bearing cells (mainly monocytes and naturalkiller [NK] cells) in two distinct ways. Eithermembrane fractalkine activated membrane frac-talkine receptor in a contact-dependent fash-ion, or the chemokine domain, released byproteolysis, could function as a soluble ligand.The transmembrane expression of fractalkineat endothelial apical surfaces suggested a meansof signaling to nearby leukocytes, and soon itwas shown that fractalkine/receptor interac-tions could arrest leukocytes under flow withoutinvolvement either of pertussis toxin-sensitiveG proteins or leukointegrins (Fong et al. 1998;Haskell et al. 1999, 2000).

Subsequent research showed that the solu-ble chemokine domain, mounted at the termi-nus of the mucin stalk, signaled by G protein–dependent interactions and mobilized calciumbut failed to activate integrins, a property incontrast to virtually all soluble chemokines.These experiments, performed nearly 20 yearsago, may be worth revisiting for guidance aswe seek to determine mechanisms by whichneuronal fractalkine signals to microglia, boththrough contact-dependent interactions and ata distance using soluble fractalkine. In additionto Gai-mediated reduction of cAMP, chemo-kine receptors signal to elevate cytosolic calci-um, activate mitogen-activated protein (MAP)kinases, and stimulate PI3K-Akt pathways. b-Arrestin recruitment and consequent receptorinternalization, at times with additional signal-ing outputs, also characterizes chemokine re-ceptors. These canonical, generic pathways pro-vide a platform for detailed characterization,which remains to be completed for practicallyall chemokine receptors in their native cellularcontexts. Only then will selective cellular re-sponses to chemokines become susceptible totherapeutic manipulation, beyond blockade oragonism.

For soluble fractalkine, structural studies in-dicate that its receptor-binding properties differ

R.M. Ransohoff and J. El Khoury

4 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

substantially from those generally reported forchemokines (Mizoue et al. 1999), suggestingthat an account of fractalkine/receptor signal-ing may well be useful for translational ap-plications. Membrane fractalkine also showeddistinct properties, as substitution of other che-mokine domains at the end of the mucin stalkdid not produce chimeric molecules that couldsupport leukocyte arrest under flow (Haskellet al. 2000). Further, the fractalkine chemokinedomain’s crystal structure showed an unusu-ally compact globular domain, suggesting amechanistic explanation for the chemokine’sdistinct functional properties (Hoover et al.2000). Outcomes of soluble versus membranefractalkine signaling modes differ in relevantdisease models of neurodegeneration. Despiteextensive descriptive cell-biology investigationof membrane and soluble fractalkine, relativelylittle is known of differential downstream sig-naling cascades.

Fractalkine/Receptor in the Nervous System

Soon after its discovery, it became clear thatfractalkine might participate in neurobiologi-cal processes. Database inspection showed thata large majority of expressed sequence tags(ESTs) for fractalkine and its receptor camefrom CNS sources, providing a rough estimateof relative tissue expression levels. Elegant insitu hybridization (Harrison et al. 1998) showeda highly provocative expression pattern. Fractal-kine was produced by neurons, with the recep-tor restricted to microglia, and it was prescientlypredicted that neuron–microglial cross talkmight be supported by this cellular distributionpattern. Surprisingly, fractalkine’s expressionpattern throughout the remainder of the bodywas starkly different, being mainly on endothe-lium. Subsequent work confirmed that fractal-kine derives from neurons in the CNS and thatthe cerebral vasculature is an outlier in its clearlack of fractalkine expression (Sunnemark et al.2005).

Within a year, reports came suggesting thatfractalkine exerted unexpected effects towardmicroglia, especially considering that chemo-kines were largely deemed inflammatory ele-

ments, which orchestrated accumulation of im-mune cells and then promoted their effectorproperties. Instead, fractalkine protected mi-croglia from Fas-mediated cell death by alter-ing the phosphorylation of apoptotic regulatorssuch as Bcl-2 and Bad (Boehme et al. 2000).Others showed that neutralizing fractalkineantibodies nearly doubled microglial produc-tion of tumor necrosis factor (TNF) in responseto lipopolysaccharide (LPS) and exerted thesame magnitude of effect on microglial toxicitytoward cocultured hippocampal neurons. In animportant follow-up experiment (Zujovic etal. 2001), this group showed in vivo that intra-cerebroventricular (ICV) installation of fractal-kine antibodies greatly increased the inflamma-tory response to ICV TNF-a. Interestingly, co-injection of fractalkine along with TNF-a hadno effect at all, strongly suggesting that tonicsoluble fractalkine signaling was not a limit-ing factor for fractalkine-mediated neuropro-tection. The cumulative interpretation of thesestudies was that fractalkine tempered microglialresponses to cardinal inflammatory stimuli.Further, without the moderating effects exertedby fractalkine, the microglial reactions to thesestimuli could potentially be neurotoxic.

Other research implied greater complex-ity; in particular, neurons in vitro and in an invivo stroke model responded to excitotoxicstimuli by increased proteolytic release of frac-talkine (Chapman et al. 2000) with no changein mRNA level. In other cells, it was shown thatproteases of the A disintegrin and metallopro-tease (ADAM) family were responsible both fortonic (Hundhausen et al. 2003) and inducible(Garton et al. 2001; Tsou et al. 2001) fractalkinerelease. Because metalloproteases were also im-plicated in fractalkine release from neurons(Chapman et al. 2000), it seems plausible thatthese same factors mediate its proteolysis fromsome neuronal cells in hippocampus and cor-tex. Subsequent work showed that cathepsin S,a distinct protease (Clark et al. 2009), was re-quired to generate soluble fractalkine from ratdorsal horn neurons.

Fractalkine signaling was also implicated inindirect neuroprotection. One particularly lu-cid series of experiments showed that neuronal

Microglia in Health and Disease

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560 5

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

fractalkine elicited adenosine release from mi-croglia, activating survival pathways in excito-toxicity-challenged neurons via adenosine A1receptors (Lauro et al. 2008, 2010). Initial find-ings in vitro were extended in vivo to a perma-nent focal ischemia model (Cipriani et al. 2011)and were enriched by showing that fractalkinesignaling also supported glutamate uptake byastrocytes (Catalano et al. 2013).

New In Vivo Tools Bring New Insights

In 2000 came the report of a genetic model forfractalkine receptor, in which the CX3CR1 cod-ing region had been replaced with GFP usinghomologous recombination, yielding mice inwhich cells transcribing the fractalkine recep-tor gene expressed GFP in the heterozygous(GFP/þ) or homozygous knockout (GFP/GFP) state (Jung et al. 2000). These mice showedno immediately evident phenotype in eitherCNS or peripheral immune responses (Junget al. 2000). As one example, the brain-stem mi-croglial morphological response to peripheralfacial nerve crush, an established model for in-ducing microgliosis (Graeber et al. 1998), wasnot affected in fractalkine receptor knockoutmice. Results were confirmed in other geneticmodels of fractalkine receptor deficiency (Has-kell et al. 2001; Tripp et al. 2001).

At about the same time, mice lacking frac-talkine were generated (Cook et al. 2001; Sor-iano et al. 2002), enabling investigators to ask ifligand and receptor-knockout mice would showcompatible phenotypes, an essential confirma-tion step for studies using these monogamousligand/receptor genetic models.

CX3CR1gfp mice showed distinct advantag-es for monitoring microglial dynamics in vivousing two-photon imaging. As described above,these mice were used to establish the remark-ably ceaseless movements of microglial process-es (Davalos et al. 2005; Nimmerjahn et al. 2005)along with their lightning-like responses to mi-croscopic foci of injury (Davalos et al. 2005).Thereafter, the concept of “resting microglia”became progressively obsolete. In initial exper-iments, mice deficient for fractalkine showedsmaller stroke volume after transient focal is-

chemia (Soriano et al. 2002), suggesting thatfractalkine/fractalkine receptor inflammatorysignaling worsened tissue damage.

Fractalkine Receptor Signaling IlluminatesRoles of Microglia in Neurodevelopment

It has become clear that parenchymal microgliaenter the CNS parenchyma early (�E10.5 inmice) during development (Ransohoff andCardona 2010) and that their proliferation andmaintenance depend in part on factors elabo-rated from neurons. After seminal documenta-tion (Tremblay et al. 2010; Schafer et al. 2012)that microglia mediate complement-dependentsynaptic pruning in some regions of the CNS, itwas pertinent to ask whether fractalkine signal-ing contributed to regulating this process, asfractalkine receptor was shown present on mi-croglial progenitors from their first entry intoparenchyma (Mizutani et al. 2012). Results ofexamining synaptic maturation in the hippo-campus of fractalkine receptor-deficient ani-mals indicated a substantial delay in CX3CR1knockouts, potentially caused by transient re-duction in microglial numbers (Paolicelli et al.2011; Ransohoff and Stevens 2011). Further,CX3CR1-deficient mice showed severely alteredsocial interaction coupled to defective networkinteractions in adulthood (Paolicelli and Gross2011; Zhan et al. 2014).

By analogy with macrophages, microgliamay carry out two major functions during de-velopment: phagocytosis and trophic-factorproduction. Phagocytosis is exemplified by syn-aptic pruning. Factor production also occursduring development and is pivotal for certainneuronal populations. Layer V cortical neuronsshow decreased survival during the first postna-tal week in fractalkine receptor deficient mice,because of reduced microglial production ofinsulin-like growth factor (IGF)-1 (Ueno et al.2013).

Microglia Play a Role in Learning-DependentSynapse Remodeling

With advancement in Cre-Lox technologies, itwas only a matter of time before the properties

R.M. Ransohoff and J. El Khoury

6 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

of CX3CR1 as a receptor expressed uniquely onmicroglia were used to target microglia and/orspecific genes in microglia to understand theirphysiological function. Using CX3CR1 CreERmice expressing tamoxifen-inducible Cre re-combinase that allow for specific expressionof the diphtheria toxin receptor in microglia,Parkhurst et al. (2013) were able to specifical-ly deplete microglia from the brain followingdiphtheria toxin administration. Mice depletedof microglia showed deficits in multiple learn-ing tasks and a significant reduction in motor-learning-dependent synapse formation. Moreinteresting is that Cre-dependent removal ofbrain-derived neurotrophic factor (BDNF) frommicroglia recapitulated one salient effect of mi-croglia depletion: impaired motor plasticity.Further, microglial BDNF increased neuronaltropomyosin-related kinase receptor B phos-phorylation, a key mediator of synaptic plastic-ity. These findings suggest that microglia mayhave important physiological functions in learn-ing and memory by promoting learning-relatedsynapse formation through BDNF signaling.A detailed discussion of the role of fractalkine-CX3CR1 signaling in synapse development andpruning can be found in Schafer and Stevens(2015).

MICROGLIA IN CNS DISORDERS

Significant advances have been made in under-standing the roles of microglia in normal brainfunctioning. In contrast, understanding the roleof microglia in CNS disorders has proven moredifficult. Brain pathology is frequently associ-ated with disruption of the blood–brain bar-rier (BBB) and recruitment of peripheral bloodmonocytes. These cells, although significantlydifferent from microglia, share several function-al and transcriptional characteristics with mi-croglia, making it more difficult to dissect theirroles from those of microglia. However, the newemerging technologies described above, whichallow specific targeting of microglial genes,could make such a task slightly easier. We willfocus here on MS and AD, two major disordersof the CNS.

Microglia in MS

It is quite possible that the first description ofmicroglia in the brain and spinal cord of a pa-tient with MS was made by Carl Froininann whoshowed that cells (likely microglia) changedtheir morphology in CNS tissues from a 22-yr-old MS patient (Froininann 1878). However,despite of decades of research, the exact role ofmicroglia in MS has not been clarified. In ani-mal models of MS, macrophages and microgliahave been reported to exert detrimental func-tions, including toxicity to neurons and oligo-dendrocyte precursor cells, release of proteases,production of inflammatory cytokines and re-active oxygen and nitrogen species, and recruit-ment and reactivation of T lymphocytes. Manystudies, however, have also reported beneficialroles of macrophages and microglia, includ-ing roles in axonal regeneration, promotion ofremyelination, clearance of inhibitory myelindebris, and the release of neurotrophic fac-tors (Rawji and Yong 2013). A main point ofconfusion is that investigators have used mi-croglia and macrophages interchangeably. Add-ing to the conceptual chaos, in EAE, a model forthe inflammatory aspects of MS (Huang et al.2006), fractalkine/receptor interactions medi-ate recruitment of disease-suppressing NK cells(Huang et al. 2006), which was shown subse-quently to be a dominant effect (Garcia et al.2013) over the effects of fractalkine/receptorinteractions on microglia. The fog may be be-ginning to dissipate, however, on the role ofmicroglia in EAE. Recently, using a CX3CR1-Cre mouse model similar to the one describedby Parkhurst et al. (2013) above, Goldmannand colleagues (2013) found that conditionaldeletion of the transforming growth factor(TGF)-b-activated kinase 1 (TAK-1) in mi-croglia only, not in neuroectodermal cells, sup-pressed EAE disease, significantly reduced CNSinflammation and diminished axonal and mye-lin damage by cell-autonomous inhibition ofthe NF-kB, JNK, and ERK-1/2 pathways. Theseresults strongly suggest that microglia may pro-mote tissue injury in EAE. Some caveats applyhowever. It is not entirely certain when theTAK-1 deletion from microglia exerts its effects.

Microglia in Health and Disease

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560 7

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Further, it remains conceivable that the tran-sient peripheral deletion of TAK-1 from allCX3CR1þ cells (an eccentricity of this geneticmodel) may play some role in the findings.

Another set of informative data came fromexamining dual-knockin mice in which Ccr2was disrupted by insertion of red fluorescentprotein (RFP), and the cross of these mice withCX3CR1gfp animals yielded animals in whichboth Ly6Chi and Ly6clo monocytes were labeled,as well as the microglia (Fig. 1). Studies at EAEonset revealed that macrophages derived fromLy6Chi monocytes initiated demyelination andmade inflammatory mediators. In contrast, mi-croglia did not associate with undisrupted axo-glial units and showed an expression profile ofrepressed metabolism (Yamasaki et al. 2014). Itwill be useful to integrate the finding from thisstudy with those in which TAK-1 was deletedfrom microglia. It is reasonable to speculate thatmicroglial activities, which promote EAE, takeplace before disease onset.

Finally, of course, the relevance of these ob-servations to MS remains to be seen.

Microglia in AD

There is definitive evidence that AD is associ-ated with significant accumulation of microgliain senile plaques (Fig. 2). Work over the pastdecade, however, suggests that, in addition tomicroglia, other mononuclear phagocytes, suchas perivascular macrophages and circulatingmonocytes, may also be involved.

Microglia are two- to fivefold more concen-trated around plaques in AD patients and mousemodels of this disease (Frautschy et al. 1998)and express major histocompatibility complexII and markers of inflammation (Tooyama etal. 1990; McGeer and McGeer 2010). b-Amy-loid (Ab) activates neonatal mouse microgliain vitro to produce reactive nitrogen speciesand TNF-a (Meda et al. 1995), a process thatis mediated in part by binding Ab to a CD36-TLR4-TLR6 receptor complex on microglia (ElKhoury et al. 2003; Stewart et al. 2010). Ab alsoactivates the NLRP3 inflammasome in microg-lia leading to neurodegeneration (Heneka et al.2013).

Although we have some insight into howmicroglia change following interaction withAb, understanding how microglia accumulateat sites of Ab deposition has proven to bemore difficult, and hinted at a role for peripheralmonocytes in AD. Transgenic mice expressingthe human Ab protein precursor with the Swe-

Figure 1. Microglia begin to clear myelin debris atEAE onset. Three-dimensional reconstructionsfrom serial block face scanning EM of spinal cordtissues of a mouse at day of EAE onset show a largeflap of myelin debris (blue) engaged by a microglialprocess (green). A nearby monocyte (red cytosol;golden nucleus) enwraps a myelin internode and isactively removing myelin. Both monocyte and mi-croglial cell (with yellow nucleus) contain myelin in-clusions (blue), although those of the monocyte arelarger and more numerous.

R.M. Ransohoff and J. El Khoury

8 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

dish mutation (APP Tg2576) and deficient in thechemokine receptor CCR2 (APP-CCR22/2)have reduced numbers of monocytes in thebrain early in the disease process, before forma-tion of senile plaques. This reduction in thenumber of monocytes was associated with in-creased mortality and higher Ab levels exclu-sively around blood vessels in the brain, suggest-ing that early monocyte accumulation promotesthe clearance of perivascular Ab and protectsmice from Ab toxicity early in the disease pro-cess (El Khoury et al. 2007). The process of Abclearance is at least, in part, mediated by phago-cytosis via the scavenger receptor SCARA1 (ElKhoury et al. 1996; Frenkel et al. 2013).

Perivascular Ab deposits are found in a highpercentage of AD patients and are a hallmark ofcerebral amyloid angiopathy. Because microgliaexpress little CCR2 (Mizutani et al. 2012), thesefindings raise the possibility that accumulationof microglia at parenchymal sites of Ab depo-sition involves a separate process that does notrequire CCR2, unlike what happens at perivas-cular sites of Ab deposition. This is indirectlysupported by results showing that selective de-pletion of perivascular macrophages exacer-bates perivascular Ab deposition, and stimula-tion of their turnover reduces perivascular load(Hawkes and McLaurin 2009). The distinctionbecomes less critical if, as proposed by Kumar-

Singh and colleagues (2005), all Ab depositsdevelop initially around blood vessels and thenthese vessels are obliterated leading to formationof the classic plaques. The latter possibility issupported by experiments in which adoptivelytransferred monocytes home to senile plaquesin a mouse model of AD (Lebson et al. 2010).

Regardless of whether Ab clearance isthe task of microglia or recruited perivascularmonocytes, the ability of microglia to clear Abappears to be reduced with disease progression.Expression of microglial Ab phagocytic recep-tors and Ab-degrading enzymes in a mousemodel of amyloid deposition in AD is signifi-cantly reduced (Hickman et al. 2008), suggest-ing that accumulation of Ab is in part the resultof a failure of microglia to adequately clear thistoxic peptide.

Additional insight into the role of microgliain AD came from analyzing the role of CX3CR1in mouse models of amyloid deposition andtauopathy. CX3CR1-deficient animals show en-hanced Ab clearance (Lee et al. 2010; Liu et al.2010), with a prominent gene-dosage effect. In-creased IL-1b is also present in the CNS of thefractalkine receptor knockout animals, and thephenotype resembles that of mice overexpress-ing IL-1b (Shaftel et al. 2007). In mice withtau pathology, absence of CX3CR1 substantiallyworsens disease progression, neuropathologi-cally, biochemically, and behaviorally (Bhaskaret al. 2010). It is plausible that increased mi-croglial production of IL-1b is responsibleboth for enhanced Ab clearance and for wors-ened tau pathology (Prinz et al. 2011).

Such findings raise the interesting ques-tion of whether soluble fractalkine (sFKN) andmembrane-tethered fractalkine (mFKN) arefunctionally distinct. The corollary questionis whether contact-dependent interactions be-tween neurons and microglia via fractalkine/re-ceptor interactions support the same functionsas does signaling at a distance through cleavedfractalkine. Studies in the periphery suggesteddistinct functions of the two-fractalkine iso-forms, as mFKN rescued survival of Ly6Clo

monocytes, whereas sFKN reversed a morpho-logical defect in CX3CR1þ gut dendritic cells(Kim et al. 2011). Use of recombinant adeno-

Figure 2. b-Amyloid (Ab) deposits are surroundedby microglia. Microglia, stained here in brown forCD11b, cluster around sites of Ab deposition, stainedin green with thioflavin S, in a mouse model of cere-bral amyloid deposition.

Microglia in Health and Disease

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560 9

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

associated virus (AAV) vectors to deliver fractal-kine isoforms showed that sFKN (but notmFKN) reversed the severely augmented toxic-ity of MPTP for substantia nigra dopamine neu-rons (Morganti et al. 2012). This observationwas extended to a therapeutic paradigm by us-ing these AAV vectors to overexpress sFKN in afractalkine-sufficient model of genetic tau pa-thology (Nash et al. 2013), with substantial ben-efit. However, the same approach was neitherbeneficial nor deleterious in an amyloid depo-sition model (Nash et al. 2013), compatible withearlier results using the LPS challenge (Zujovicet al. 2001).

Because recent findings suggest that tau pa-thology may occur following Ab deposition,and based on the above discussion, we proposethat the role of microglia in AD evolves withdisease progression. Microglia initially interactwith Ab possibly in an attempt to clear it. Butthe continuous accumulation of Ab also leads toinduction of inflammatory cytokines that re-duce the Ab clearance ability of microglia andlead to tau pathology, initiating a self-perpetu-ating loop that culminates in worsening disease(see Table 1). This is reminiscent of the sequenceof events that occur in atherosclerosis, anotherdegenerative disease of aging. Monocytes areinitially recruited to sites of deposition of mod-ified lipoproteins, likely in an attempt to clearthem. Continued accumulation of such lipo-proteins overwhelms the ability of the mono-cytes to clear them and transforms these cellsinto foam cells that contribute to disease prog-ression. Of interest, scavenger receptors thatappear to mediate the interactions with Ab in

AD also mediate the interactions of monocyteswith modified lipoproteins in atherosclerosis.

In addition to their roles in MS and AD,microglia have been implicated in a numberof neurological and neuropsychiatric disorders(El Khoury 2010). For example, microglia ina mouse model of amyotrophic lateral sclerosis(ALS) have a distinct molecular signature (Chiuet al. 2013). A defect in the microglial/myeloidgene Hoxb8 is associated with compulsivegrooming similar to behavior in humans withobsessive–compulsive disorder (Chen et al.2010). Another example is Rett syndrome, anX-linked autism spectrum disorder. In a mousemodel of Rett syndrome, normal microgliacan arrest the pathology and attenuate diseasesymptoms (Derecki et al. 2012).

TOWARD A MOLECULAR DEFINITIONOF MICROGLIA

It is evident from the above discussion thatthe ability to distinguish microglia from othercells of the CNS as well as from other mononu-clear phagocytes is crucial to understanding thefunctions of these cells in physiological andpathological processes. Development of meth-ods to isolate microglia from adult animals withhigh purity and yield as well as recent advancesin approaches to define the transcriptomes ofvarious cells by RNASeq and microarrays al-lowed four independent groups to define thetranscriptomes of adult mouse microglia andmake these data publicly available in 2013(Beutner et al. 2013; Chiu et al. 2013; Hickmanet al. 2013; Butovsky et al. 2014). These resultsshowed that, although microglia share severalimportant transcripts with other mononuclearphagocytes, they also have significant differenc-es. Analysis of these datasets shows that P2ry12and P2ry13, Tmem119, Gpr34, Siglech, Trem2,and CX3CR1 are among the transcripts unique-ly expressed in microglia compared with oth-er mononuclear phagocytes. Macrophage-en-riched genes include fibronectin, the chemokineCxcl13, and the endothelin B receptor.

Analysis of these datasets also allowedcorrelation between microglial gene expressionand function. For example, a cluster of genes

Table 1. The neuroimmune system plays beneficialand detrimental roles in Alzheimer’s disease

Potential beneficial roles

Potential detrimental

roles

Ab phagocytosisRelease of Ab-degrading

enzymesClearance of debris and

degenerating neuronsRelease of neurotropic

factors

Release of inflammatorycytokines

Release of reactiveoxygen and nitrogenspecies

R.M. Ransohoff and J. El Khoury

10 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

termed “sensome” was introduced that definesthe genes used by microglial to sense their en-vironment (Hickman et al. 2013), a major func-tion of microglia described using two-photonimaging (Davalos et al. 2005; Nimmerjahn et al.2005). This cluster includes 100 genes that con-stitute the microglial toolset for sensing changesin the brain’s milieu and define the apparatusthat microglia use to perform various homeo-static functions including sensing of chemo-kines and cytokines, purinergic molecules, in-organic substances, changes in pH and aminoacids. Defining the microglial sensome underphysiological conditions, establishes a baselineto which we can compare and identify changesthat occur in this sensome under pathologi-cal conditions. Highlighting the importance ofthese datasets, two members of the microglialsensome, TREM2 and CD33, have been foundto be independent risk factors for late onset AD(Jonsson et al. 2012; Bradshaw et al. 2013; Guer-reiro et al. 2013).

Another important result in these datasetsis the finding that resident microglia expresshigh levels of several antimicrobial peptidesnot previously known to be expressed on thesecells (Hickman et al. 2013). These includeCamp, the cathelicidin-related antimicrobialpeptide and Ngp, the neutrophilic granule pro-tein. The high level of expression of these pep-tides indicates a high level of readiness by thenormal “quiescent” resident microglia to per-form its innate host defense function in the ab-sence of adaptive immune molecules and cells.These findings further support the concept thatthere is no “resting” microglia, and that thesecells are highly dynamic cells important for sur-veillance, protection, and housekeeping of theneural milieu.

CONCLUSIONS

It is an exciting time to study microglia. Basedon the rapid pace of discovery in the past de-cade, and the continued development of newtechnologies, it is highly likely that the nextdecade will bring our level of understandingof these cells on par to our levels of understand-ing of other innate immune cells in the periph-

ery and allow us to harness such knowledge todefine the role of these cells in normal and path-ological processes and to possibly target thesecells for diagnosis and/or treatment of variousCNS disorders. To fully define the exact roles ofmicroglia in these disorders, experiments map-ping the gene expression profiles of microgliaand other neuroimmune cells in mouse modelsand patient samples and at multiple stages ofvarious CNS disorders need to be performed(El Khoury 2010).

REFERENCES�Reference is also in this collection.

Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FM. 2007.Local self-renewal can sustain CNS microglia mainte-nance and function throughout adult life. Nat Neurosci10: 1538–1543.

Alliot F, Lecain E, Grima B, Pessac B. 1991. Microglial pro-genitors with a high proliferative potential in the embry-onic and adult mouse brain. Proc Natl Acad Sci 88: 1541–1545.

Alliot F, Godin I, Pessac B. 1999. Microglia derive from pro-genitors, originating from the yolk sac, and which prolif-erate in the brain. Brain Res Dev Brain Res 117: 145–152.

Bazan JF, Bacon KB, Hardiman G, Wang W, Soo K, Rossi D,Greaves DR, Zlotnik A, Schall TJ. 1997. A new class ofmembrane-bound chemokine with a CX3C motif. Nature385: 640–644.

Beutner C, Linnartz-Gerlach B, Schmidt SV, Beyer M, Mall-mann MR, Staratschek-Jox A, Schultze JL, Neumann H.2013. Unique transcriptome signature of mouse microg-lia. Glia 61: 1429–1442.

Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A,Ransohoff RM, Lamb BT. 2010. Regulation of tau pathol-ogy by the microglial fractalkine receptor. Neuron 68:19–31.

Boehme SA, Lio FM, Maciejewski-Lenoir D, Bacon KB,Conlon PJ. 2000. The chemokine fractalkine inhibitsFas-mediated cell death of brain microglia. J Immunol165: 397–403.

Bradshaw EM, Chibnik LB, Keenan BT, Ottoboni L, Raj T,Tang A, Rosenkrantz LL, Imboywa S, Lee M, Von Korff A,et al. 2013. CD33 Alzheimer’s disease locus: Alteredmonocyte function and amyloid biology. Nat Neurosci16: 848–850.

Butovsky O, Jedrychowski MP, Moore CS, Cialic R, LanserAJ, Gabriely G, Koeglsperger T, Dake B, Wu PM, DoykanCE, et al. 2014. Identification of a unique TGF-b-depen-dent molecular and functional signature in microglia.Nat Neurosci 17: 131–143.

Cardona AE, Sasse ME, Liu L, Cardona SM, Mizutani M,Savarin C, Hu T, Ransohoff RM. 2008. Scavenging roles ofchemokine receptors: Chemokine receptor deficiency isassociated with increased levels of ligand in circulationand tissues. Blood 112: 256–263.

Microglia in Health and Disease

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560 11

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Catalano M, Lauro C, Cipriani R, Chece G, Ponzetta A, DiAngelantonio S, Ragozzino D, Limatola C. 2013.CX3CL1 protects neurons against excitotoxicity enhanc-ing GLT-1 activity on astrocytes. J Neuroimmunol 263:75–82.

Chapman GA, Moores K, Harrison D, Campbell CA, Stew-art BR, Strijbos PJ. 2000. Fractalkine cleavage from neu-ronal membranes represents an acute event in the inflam-matory response to excitotoxic brain damage. J Neurosci20: RC87.

Charo IF, Ransohoff RM. 2006. The many roles of chemo-kines and chemokine receptors in inflammation. N EnglJ Med 354: 610–621.

Chen SK, Tvrdik P, Peden E, Cho S, Wu S, Spangrude G,Capecchi MR. 2010. Hematopoietic origin of patholog-ical grooming in Hoxb8 mutant mice. Cell 141: 775–785.

Chiu IM, Morimoto ET, Goodarzi H, Liao JT, O’Keeffe S,Phatnani HP, Muratet M, Carroll MC, Levy S, Tavazoie S,et al. 2013. A neurodegeneration-specific gene-expres-sion signature of acutely isolated microglia from anamyotrophic lateral sclerosis mouse model. Cell Rep 4:385–401.

Cipriani R, Villa P, Chece G, Lauro C, Paladini A, Micotti E,Perego C, De Simoni MG, Fredholm BB, Eusebi F, et al.2011. CX3CL1 is neuroprotective in permanent focal ce-rebral ischemia in rodents. J Neurosci 31: 16327–16335.

Clark AK, Yip PK, Malcangio M. 2009. The liberation offractalkine in the dorsal horn requires microglial cathep-sin S. J Neurosci 29: 6945–6954.

Cook DN, Chen SC, Sullivan LM, Manfra DJ, WiekowskiMT, Prosser DM, Vassileva G, Lira SA. 2001. Generationand analysis of mice lacking the chemokine fractalkine.Mol Cell Biol 21: 3159–3165.

Cuzner ML, Hayes GM, Newcombe J, Woodroofe MN. 1988.The nature of inflammatory components during demye-lination in multiple sclerosis. J Neuroimmunol 20: 203–209.

Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S,Littman DR, Dustin ML, Gan WB. 2005. ATP mediatesrapid microglial response to local brain injury in vivo.Nat Neurosci 8: 752–758.

del Rıo-Hortega Pd. 1918. Noticia de un nuevo y facil me-todo para la coloracion de la neuroglia y el tejido con-juntivo [A new and easiest approach for staining glia andconnective tissue]. Trab Lab Inv Biol 15: 367–378.

del Rıo-Hortega Pd. 1919. El “tercer elemento” de los cen-tros nerviosos. Poder fagocitario y mivilidad de la micro-glıa [The “third element” of the central nervous system.Phagocytic power of microglia]. Bol Soc Esp Biol 8: 62–82.

del Rıo-Hortega Pd. 1920. La microglıa y su transformacionen celulas en bastoncito y cuerpos granuloadiposos[Transformation in the microglial cell bodies]. Trab LabInv Biol 18: 37–82.

del Rıo-Hortega Pd. 1921. Histogenesis y evolucion normal,exodo y distrubucion regional de la microglıa [Evolutionand regional distribution of microglia]. Mem Real Soc EspHist Nat 11: 213–268.

Derecki NC, Cronk JC, Lu Z, Xu E, Abbott SB, Guyenet PG,Kipnis J. 2012. Wild-type microglia arrest pathology in amouse model of Rett syndrome. Nature 484: 105–109.

El Khoury J. 2010. Neurodegeneration and the neuroim-mune system. Nat Med 16: 1369–1370.

El Khoury J, Hickman SE, Thomas CA, Cao L, SilversteinSC, Loike JD. 1996. Scavenger receptor-mediated adhe-sion of microglia to b-amyloid fibrils. Nature 382: 716–719.

El Khoury JB, Moore KJ, Means TK, Leung J, Terada K, ToftM, Freeman MW, Luster AD. 2003. CD36 mediates theinnate host response tob-amyloid. J Exp Med 197: 1657–1666.

El Khoury J, Toft M, Hickman SE, Means TK, Terada K,Geula C, Luster AD. 2007. Ccr2 deficiency impairs mi-croglial accumulation and accelerates progression of Alz-heimer-like disease. Nat Med 13: 432–438.

Fong AM, Robinson LA, Steeber DA, Tedder TF, Yoshie O,Imai T, Patel DD. 1998. Fractalkine and CX3CR1 mediatea novel mechanism of leukocyte capture, firm adhesion,and activation under physiologic flow. J Exp Med 188:1413–1419.

Frautschy SA, Yang F, Irrizarry M, Hyman B, Saido TC,Hsiao K, Cole GM. 1998. Microglial response to amyloidplaques in APPsw transgenic mice. Am J Pathol 152: 307–317.

Frenkel D, Wilkinson K, Zhao L, Hickman SE, Means TK,Puckett L, Farfara D, Kingery ND, Weiner HL, El KhouryJ. 2013. Scara1 deficiency impairs clearance of solubleamyloid-b by mononuclear phagocytes and acceleratesAlzheimer’s-like disease progression. Nat Commun 4:2030.

Froininann C. 1878. Untersuchungen uber die Gewebveran-derungen bei der multiplen sklerose des gehirns und rucken-marks [Studies in multiple sclerosis of the brain]. GustavFischer, Jena, Germany.

Garcia JA, Pino PA, Mizutani M, Cardona SM, Charo IF,Ransohoff RM, Forsthuber TG, Cardona AE. 2013. Reg-ulation of adaptive immunity by the fractalkine recep-tor during autoimmune inflammation. J Immunol 191:1063–1072.

Garton KJ, Gough PJ, Blobel CP, Murphy G, Greaves DR,Dempsey PJ, Raines EW. 2001. Tumor necrosis factor-a-converting enzyme (ADAM17) mediates the cleavageand shedding of fractalkine (CX3CL1). J Biol Chem 276:37993–38001.

Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S,Mehler MF, Conway SJ, Ng LG, Stanley ER, et al. 2010.Fate mapping analysis reveals that adult microglia derivefrom primitive macrophages. Science 330: 841–845.

Giulian D, Baker TJ. 1986. Characterization of ameboidmicroglia isolated from developing mammalian brain.J Neurosci 6: 2163–2178.

Goldmann T, Wieghofer P, Muller PF, Wolf Y, Varol D, YonaS, Brendecke SM, Kierdorf K, Staszewski O, Datta M,et al. 2013. A new type of microglia gene targeting showsTAK1 to be pivotal in CNS autoimmune inflammation.Nat Neurosci 16: 1618–1626.

Graeber MB, Lopez-Redondo F, Ikoma E, Ishikawa M, ImaiY, Nakajima K, Kreutzberg GW, Kohsaka S. 1998. Themicroglia/macrophage response in the neonatal rat facialnucleus following axotomy. Brain Res 813: 241–253.

Greter M, Lelios I, Pelczar P, Hoeffel G, Price J, Leboeuf M,Kundig TM, Frei K, Ginhoux F, Merad M, et al. 2012.Stroma-derived interleukin-34 controls the development

R.M. Ransohoff and J. El Khoury

12 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

and maintenance of Langerhans cells and the mainte-nance of microglia. Immunity 37: 1050–1060.

Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E,Majounie E, Cruchaga C, Sassi C, Kauwe JS, Younkin S,et al. 2013. TREM2 variants in Alzheimer’s disease. NEngl J Med 368: 117–127.

Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, McNa-mara RK, Streit WJ, Salafranca MN, Adhikari S, Thomp-son DA, et al. 1998. Role for neuronally derived fractal-kine in mediating interactions between neurons andCX3CR1-expressing microglia. Proc Natl Acad Sci 95:10896–10901.

Haskell CA, Cleary MD, Charo IF. 1999. Molecular uncou-pling of fractalkine-mediated cell adhesion and signaltransduction. Rapid flow arrest of CX3CR1-express-ing cells is independent of G-protein activation. J BiolChem 274: 10053–10058.

Haskell CA, Cleary MD, Charo IF. 2000. Unique role of thechemokine domain of fractalkine in cell capture. Kineticsof receptor dissociation correlate with cell adhesion. J BiolChem 275: 34183–34189.

Haskell CA, Hancock WW, Salant DJ, Gao W, Csizmadia V,Peters W, Faia K, Fituri O, Rottman JB, Charo IF. 2001.Targeted deletion of CX3CR1 reveals a role for fractalkinein cardiac allograft rejection. J Clin Invest 108: 679–688.

Hawkes CA, McLaurin J. 2009. Selective targeting of peri-vascular macrophages for clearance of b-amyloid in ce-rebral amyloid angiopathy. Proc Natl Acad Sci 106: 1261–1266.

Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME,Gan WB, Julius D. 2006. The P2Y12 receptor regulatesmicroglial activation by extracellular nucleotides. NatNeurosci 9: 1512–1519.

Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S,Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC,et al. 2013. NLRP3 is activated in Alzheimer’s disease andcontributes to pathology in APP/PS1 mice. Nature 493:674–678.

Hickman SE, Allison EK, El Khoury J. 2008. Microglial dys-function and defective b-amyloid clearance pathways inaging Alzheimer’s disease mice. J Neurosci 28: 8354–8360.

Hickman SE, Kingery ND, Ohsumi TK, Borowsky ML,Wang LC, Means TK, El Khoury J. 2013. The microglialsensome revealed by direct RNA sequencing. Nat Neuro-sci 16: 1896–1905.

Hoover DM, Mizoue LS, Handel TM, Lubkowski J. 2000.The crystal structure of the chemokine domain of frac-talkine shows a novel quaternary arrangement. J BiolChem 275: 23187–23193.

Horuk R. 1994. Molecular properties of the chemokine re-ceptor family. Trends Pharmacol Sci 15: 159–165.

Huang D, Shi FD, Jung S, Pien GC, Wang J, Salazar-MatherTP, He TT, Weaver JT, Ljunggren HG, Biron CA, et al.2006. The neuronal chemokine CX3CL1/fractalkine se-lectively recruits NK cells that modify experimental au-toimmune encephalomyelitis within the central nervoussystem. FASEB J 20: 896–905.

Hume DA, Perry VH, Gordon S. 1983. Immunohistochem-ical localization of a macrophage-specific antigen in de-veloping mouse retina: Phagocytosis of dying neurons

and differentiation of microglial cells to form a regulararray in the plexiform layers. J Cell Biol 97: 253–257.

Hundhausen C, Misztela D, Berkhout TA, Broadway N, Saf-tig P, Reiss K, Hartmann D, Fahrenholz F, Postina R,Matthews V, et al. 2003. The disintegrin-like metallopro-teinase ADAM10 is involved in constitutive cleavage ofCX3CL1 (fractalkine) and regulates CX3CL1-mediatedcell–cell adhesion. Blood 102: 1186–1195.

Imai Y, Ibata I, Ito D, Ohsawa K, Kohsaka S. 1996. A novelgene iba1 in the major histocompatibility complex classIII region encoding an EF hand protein expressed in amonocytic lineage. Biochem Biophys Res Commun 224:855–862.

Jonsson T, Stefansson H, Steinberg S, Jonsdottir I, JonssonPV, Snaedal J, Bjornsson S, Huttenlocher J, Levey AI, LahJJ, et al. 2012. Variant of TREM2 associated with the riskof Alzheimer’s disease. N Engl J Med 368: 107–116.

Jung S, Aliberti J, Graemmel P, Sunshine MJ, KreutzbergGW, Sher A, Littman DR. 2000. Analysis of fractalkinereceptor CX3CR1 function by targeted deletion and greenfluorescent protein reporter gene insertion. Mol Cell Biol20: 4106–4114.

Kierdorf K, Prinz M. 2013. Factors regulating microglia ac-tivation. Front Cell Neurosci 7: 44.

Kim KW, Vallon-Eberhard A, Zigmond E, Farache J, ShezenE, Shakhar G, Ludwig A, Lira SA, Jung S. 2011. In vivostructure/function and expression analysis of the CX3Cchemokine fractalkine. Blood 118: e156–e167.

Kumar-Singh S, Pirici D, McGowan E, Serneels S, CeuterickC, Hardy J, Duff K, Dickson D, Van Broeckhoven C. 2005.Dense-core plaques in Tg2576 and PSAPP mouse modelsof Alzheimer’s disease are centered on vessel walls. Am JPathol 167: 527–543.

Lauro C, Di Angelantonio S, Cipriani R, Sobrero F, AntonilliL, Brusadin V, Ragozzino D, Limatola C. 2008. Activity ofadenosine receptors type 1 is required for CX3CL1-me-diated neuroprotection and neuromodulation in hippo-campal neurons. J Immunol 180: 7590–7596.

Lauro C, Cipriani R, Catalano M, Trettel F, Chece G, Brusa-din V, Antonilli L, van Rooijen N, Eusebi F, Fredholm BB,et al. 2010. Adenosine A1 receptors and microglial cellsmediate CX3CL1-induced protection of hippocampalneurons against Glu-induced death. Neuropsychophar-macology 35: 1550–1559.

Lebson L, Nash K, Kamath S, Herber D, Carty N, Lee DC, LiQ, Szekeres K, Jinwal U, Koren J, et al. 2010. TraffickingCD11b-positive blood cells deliver therapeutic genes tothe brain of amyloid-depositing transgenic mice. J Neu-rosci 30: 9651–9658.

Lee S, Varvel NH, Konerth ME, Xu G, Cardona AE, Ranso-hoff RM, Lamb BT. 2010. CX3CR1 deficiency altersmicroglial activation and reduces b-amyloid depositionin two Alzheimer’s disease mouse models. Am J Pathol177: 2549–2562.

Liu Z, Condello C, Schain A, Harb R, Grutzendler J. 2010.CX3CR1 in microglia regulates brain amyloid depositionthrough selective protofibrillar amyloid-b phagocytosis.J Neurosci 30: 17091–17101.

Ma Q, Jones D, Borghesani PR, Segal RA, Nagasawa T, Ki-shimoto T, Bronson RT, Springer TA. 1998. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar

Microglia in Health and Disease

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560 13

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

neuron migration in CXCR4 or SDF-1 deficient mice.Proc Natl Acad Sci 95: 9448–9453.

McGeer EG, McGeer PL. 2010. Neuroinflammation in Alz-heimer’s disease and mild cognitive impairment: A fieldin its infancy. J Alzheimers Dis 19: 355–361.

McGeer PL, Itagaki S, Tago H, McGeer EG. 1987. Reactivemicroglia in patients with senile dementia of the Alz-heimer type are positive for the histocompatibility gly-coprotein HLA-DR. Neurosci Lett 79: 195–200.

McKercher SR, Torbett BE, Anderson KL, Henkel GW, VestalDJ, Baribault H, Klemsz M, Feeney AJ, Wu GE, Paige CJ,et al. 1996. Targeted disruption of the PU.1 gene resultsin multiple hematopoietic abnormalities. EMBO J 15:5647–5658.

Meda L, Cassatella MA, Szendrei GI, Otvos L Jr, Baron P,Villalba M, Ferrari D, Rossi F. 1995. Activation of micro-glial cells by b-amyloid protein and interferon-g. Nature374: 647–650.

Mizoue LS, Bazan JF, Johnson EC, Handel TM. 1999. So-lution structure and dynamics of the CX3C chemokinedomain of fractalkine and its interaction with an N-ter-minal fragment of CX3CR1. Biochemistry 38: 1402–1414.

Mizutani M, Pino PA, Saederup N, Charo IF, Ransohoff RM,Cardona AE. 2012. The fractalkine receptor but notCCR2 is present on microglia from embryonic develop-ment throughout adulthood. J Immunol 188: 29–36.

Morganti JM, Nash KR, Grimmig BA, Ranjit S, Small B,Bickford PC, Gemma C. 2012. The soluble isoform ofCX3CL1 is necessary for neuroprotection in a mousemodel of Parkinson’s disease. J Neurosci 32: 14592–14601.

Nakayama T, Watanabe Y, Osio N, Higuchi T, Shigeta A,Mizuguchi N, Katou F, Hashimoto K, Kawada A, YoshieO. 2010. Eotaxin-3/CC chemokine ligand 26 is a func-tional ligand for CX3CR1. J Immunol 185: 6472–6479.

Nash KR, Lee DC, Hunt JB Jr, Morganti JM, Selenica ML,Moran P, Reid P, Brownlow M, Guang-Yu Yang C, SavaliaM, et al. 2013. Fractalkine overexpression suppressestau pathology in a mouse model of tauopathy. NeurobiolAging 34: 1540–1548.

Nimmerjahn A, Kirchhoff F, Helmchen F. 2005. Resting mi-croglial cells are highly dynamic surveillants of brain pa-renchyma in vivo. Science 308: 1314–1318.

Paolicelli RC, Gross CT. 2011. Microglia in development:Linking brain wiring to brain environment. NeuronGlia Biol 7: 77–83.

Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M,Panzanelli P, Giustetto M, Ferreira TA, Guiducci E, Du-mas L, et al. 2011. Synaptic pruning by microglia is nec-essary for normal brain development. Science 333: 1456–1458.

Parkhurst CN, Yang G, Ninan I, Savas JN, Yates JR III, La-faille JJ, Hempstead BL, Littman DR, Gan WB. 2013.Microglia promote learning-dependent synapse forma-tion through brain-derived neurotrophic factor. Cell 155:1596–1609.

Perry VH, Hume DA, Gordon S. 1985. Immunohisto-chemical localization of macrophages and microglia inthe adult and developing mouse brain. Neuroscience 15:313–326.

Prinz M, Priller J, Sisodia SS, Ransohoff RM. 2011. Hetero-geneity of CNS myeloid cells and their roles in neuro-degeneration. Nat Neurosci 14: 1227–1235.

Ransohoff RM. 2005. Selective leukocyte chemoattractantsemerge from the primeval sup(ernatants). J Immunol175: 5567–5568.

Ransohoff RM, Cardona AE. 2010. The myeloid cells of thecentral nervous system parenchyma. Nature 468: 253–262.

Ransohoff RM, Stevens B. 2011. Neuroscience. How manycell types does it take to wire a brain? Science 333: 1391–1392.

Rawji KS, Yong VW. 2013. The benefits and detriments ofmacrophages/microglia in models of multiple sclerosis.Clin Dev Immunol 2013: 948976.

Rot A, von Andrian UH. 2004. Chemokines in innate andadaptive host defense: Basic chemokinese grammar forimmune cells. Annu Rev Immunol 22: 891–928.

Rozemuller JM, Eikelenboom P, Stam FC. 1986. Role ofmicroglia in plaque formation in senile dementia of theAlzheimer type. An immunohistochemical study. Vir-chows Arch B Cell Pathol Incl Mol Pathol 51: 247–254.

� Schafer DP, Stevens B. 2015. Microglia function in centralnervous system development and plasticity. Cold SpringHarb Perspect Biol doi: 10.1101/cshperspect.a020545.

Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mar-dinly AR, Yamasaki R, Ransohoff RM, Greenberg ME,Barres BA, Stevens B. 2012. Microglia sculpt postnatalneural circuits in an activity and complement-dependentmanner. Neuron 74: 1–15.

Schulz C, Gomez Perdiguero E, Chorro L, Szabo-Rogers H,Cagnard N, Kierdorf K, Prinz M, Wu B, Jacobsen SE,Pollard JW, et al. 2012. A lineage of myeloid cells inde-pendent of Myb and hematopoietic stem cells. Science336: 86–90.

Shaftel SS, Kyrkanides S, Olschowka JA, Miller JN, JohnsonRE, O’Banion MK. 2007. Sustained hippocampal IL-1boverexpression mediates chronic neuroinflammation andameliorates Alzheimer plaque pathology. J Clin Invest117: 1595–1604.

Soriano SG, Amaravadi LS, Wang YF, Zhou H, Yu GX, TonraJR, Fairchild-Huntress V, Fang Q, Dunmore JH, HuszarD, et al. 2002. Mice deficient in fractalkine are less sus-ceptible to cerebral ischemia-reperfusion injury. J Neuro-immunol 125: 59–65.

Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J,Halle A, Rayner KJ, Boyer L, Zhong R, Frazier WA, et al.2010. CD36 ligands promote sterile inflammationthrough assembly of a Toll-like receptor 4 and 6 hetero-dimer. Nat Immunol 11: 155–161.

Sunnemark D, Eltayeb S, Nilsson M, Wallstrom E, LassmannH, Olsson T, Berg AL, Ericsson-Dahlstrand A. 2005.CX3CL1 (fractalkine) and CX3CR1 expression in myelinoligodendrocyte glycoprotein-induced experimental au-toimmune encephalomyelitis: Kinetics and cellular ori-gin. J Neuroinflammation 2: 17.

Tooyama I, Kimura H, Akiyama H, McGeer PL. 1990. Re-active microglia express class I and class II major histo-compatibility complex antigens in Alzheimer’s disease.Brain Res 523: 273–280.

R.M. Ransohoff and J. El Khoury

14 Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

Tremblay ME, Lowery RL, Majewska AK. 2010. Microglialinteractions with synapses are modulated by visual expe-rience. PLoS Biol 8: e1000527.

Tripp RA, Jones LP, Haynes LM, Zheng H, Murphy PM,Anderson LJ. 2001. CX3C chemokine mimicry by respi-ratory syncytial virus G glycoprotein. Nat Immunol 2:732–738.

Tsou CL, Haskell CA, Charo IF. 2001. Tumor necrosis factor-a-converting enzyme mediates the inducible cleavage offractalkine. J Biol Chem 276: 44622–44626.

Ueno M, Fujita Y, Tanaka T, Nakamura Y, Kikuta J, Ishii M,Yamashita T. 2013. Layer V cortical neurons require mi-croglial support for survival during postnatal develop-ment. Nat Neurosci 16: 543–551.

Walz W, Ilschner S, Ohlemeyer C, Banati R, Kettenmann H.1993. Extracellular ATP activates a cation conductanceand a Kþ conductance in cultured microglial cells frommouse brain. J Neurosci 13: 4403–4411.

Wang Y, Szretter KJ, Vermi W, Gilfillan S, Rossini C, Cella M,Barrow AD, Diamond MS, Colonna M. 2012. IL-34 is atissue-restricted ligand of CSF1R required for the devel-opment of Langerhans cells and microglia. Nat Immunol13: 753–760.

Yamasaki R, Lu H, Butovsky O, Ohno N, Rietsch AM, CialicR, Wu PM, Doykan CE, Lin J, Cotleur AC, et al. 2014.Differential roles of microglia and monocytes in the in-flamed central nervous system. J Exp Med 211: 1533–1549.

Zhan Y, Paolicelli RC, Sforazzini F, Weinhard L, Bolasco G,Pagani F, Vyssotski AL, Bifone A, Gozzi A, Ragozzino D,et al. 2014. Deficient neuron-microglia signaling resultsin impaired functional brain connectivity and social be-havior. Nat Neurosci 17: 400–406.

Zou YR, Kottmann AH, Kuroda M, Taniuchi I, Littman DR.1998. Function of the chemokine receptor CXCR4 inhaematopoiesis and in cerebellar development. Nature393: 595–599.

Zucker-Franklin D, Warfel A, Grusky G, Frangione B, TeitelD. 1987. Novel monocyte-like properties of microglial/astroglial cells. Constitutive secretion of lysozyme andcystatin-C. Lab Invest 57: 176–185.

Zujovic V, Schussler N, Jourdain D, Duverger D, Taupin V.2001. In vivo neutralization of endogenous brain fractal-kine increases hippocampal TNF-a and 8-isoprostaneproduction induced by intracerebroventricular injectionof LPS. J Neuroimmunol 115: 135–143.

Microglia in Health and Disease

Cite this article as Cold Spring Harb Perspect Biol 2016;8:a020560 15

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from

September 9, 20152016; doi: 10.1101/cshperspect.a020560 originally published onlineCold Spring Harb Perspect Biol 

 Richard M. Ransohoff and Joseph El Khoury Microglia in Health and Disease

Subject Collection Glia

MaintenanceThe Nodes of Ranvier: Molecular Assembly and

Matthew N. Rasband and Elior Peles

Oligodendrocyte Development and PlasticityDwight E. Bergles and William D. Richardson

Microglia in Health and DiseaseRichard M. Ransohoff and Joseph El Khoury Support

Oligodendrocytes: Myelination and Axonal

Mikael Simons and Klaus-Armin NaveThe Astrocyte: Powerhouse and Recycling Center

Bruno Weber and L. Felipe Barros Central Nervous System GliaDrosophila

Marc R. Freeman

Development and PlasticityMicroglia Function in Central Nervous System

Dorothy P. Schafer and Beth StevensSynapse: Adaptable, Multitasking Glial CellsPerisynaptic Schwann Cells at the Neuromuscular

Chien-Ping Ko and Richard Robitaille

the Central Nervous SystemOligodendrocyte Development and Myelination in Transcriptional and Epigenetic Regulation of

Ben Emery and Q. Richard Lu

and EliminationAstrocytes Control Synapse Formation, Function,

Won-Suk Chung, Nicola J. Allen and Cagla Eroglu

ControversiesOrigin of Microglia: Current Concepts and Past

Florent Ginhoux and Marco Prinz

Schwann Cell MyelinationJames L. Salzer

Remyelination−−Glia Disease and RepairRobin J.M. Franklin and Steven A. Goldman Repair

Schwann Cells: Development and Role in Nerve

LloydKristján R. Jessen, Rhona Mirsky and Alison C.

Astrocytes in Neurodegenerative DiseaseHemali Phatnani and Tom Maniatis

Perineurial GliaSarah Kucenas

http://cshperspectives.cshlp.org/cgi/collection/ For additional articles in this collection, see

Copyright © 2016 Cold Spring Harbor Laboratory Press; all rights reserved

on September 26, 2018 - Published by Cold Spring Harbor Laboratory Press http://cshperspectives.cshlp.org/Downloaded from